Download as pdf or txt
Download as pdf or txt
You are on page 1of 8

254 Exp Physiol 93.

2 pp 254–261

Experimental Physiology

Magnesium sulphate treatment decreases blood–brain


barrier permeability during acute hypertension in
pregnant rats
Anna G. Euser, Lisa Bullinger and Marilyn J. Cipolla
Departments of Neurology, Obstetrics and Gynecology, and Pharmacology, University of Vermont College of Medicine, Burlington, VT 05405, USA

Eclampsia is associated with increased blood–brain barrier (BBB) permeability and formation of
cerebral oedema. Magnesium sulphate is used to treat eclampsia despite an unclear mechanism of
action. This study was to determine the effect of magnesium sulphate on in vivo BBB permeability
and formation of cerebral oedema during acute hypertension and on brain aquaporin-4 (AQP4)
protein expression. An in vivo model of hypertensive encephalopathy was used in late-pregnant
(LP) rats following magnesium sulphate treatment, 270 mg kg−1 I.P. injection every 4 h for 24 h.
Permeability of the BBB was determined by in situ brain perfusion of Evan’s Blue (EB) and sodium
fluorescein (NaFl), and dye clearance determined by fluorescence spectrophotometry. Cerebral
oedema was determined following acute hypertension by measuring brain water content. The
effect of magnesium treatment on AQP4 expression was determined by Western blot analysis.
Acute hypertension with autoregulatory breakthrough increased BBB permeability to EB in both
brain regions studied (P < 0.05). Magnesium attenuated BBB permeability to EB during acute
hypertension by 41% in the posterior cerebrum (P < 0.05) but had no effect in the anterior
cerebrum (P > 0.05). Treatment with magnesium did not change NaFl permeability, cerebral
oedema formation or AQP4 expression. In summary, BBB permeability to Evan’s Blue was
increased by acute hypertension in LP rats, and this was attenuated by treatment with magnesium
sulphate. The greatest effect on BBB permeability to EB was in the posterior cerebrum, an area
particularly susceptible to oedema formation during eclampsia.
(Received 15 August 2007; accepted after revision 2 October 2007; first published online 12 October 2007)
Corresponding author M. J. Cipolla: University of Vermont, Department of Neurology, 89 Beaumont Avenue, Given
C454, Burlington, VT 05405, USA. Email: marilyn.cipolla@uvm.edu

Eclampsia is a serious hypertensive disorder of pregnancy promotes oedema formation under conditions of acute
associated with increased blood–brain barrier (BBB) hypertension.
permeability and subsequent vasogenic oedema formation Magnesium sulphate is widely used in North America
(Schwartz et al. 1992, 2000; Engelter et al. 2000; Zeeman both to prevent and to treat eclamptic convulsions
et al. 2004). This condition is thought to be a form of (Witlin & Sibai, 1998). This treatment has been proven
hypertensive encephalopathy (HTE; Schwartz et al. 1992, to be more effective than anticonvulsant drugs and
2000; Easton, 1998), and both eclampsia and HTE are placebo (The Eclampsia Trial Collaborative Group, 1995;
causes of posterior reversible encephalopathy syndrome Altman et al. 2002), though the mechanism of action
(PRES; Hinchey et al. 1996; Lamy et al. 2004). Using a remains unclear. Some studies suggest that magnesium
model of HTE, we previously showed that acutely elevated may prevent eclamptic seizures via vasodilatation in
arterial pressure with breakthrough of cerebral blood the cerebral circulation (Belfort & Moise, 1992; Belfort
flow (CBF) autoregulation caused a significant increase et al. 1993; Naidu et al. 1996). However, magnesium
in cerebral oedema formation in late-pregnant (LP) rats, treatment has also been reported to have little to no
which was not seen in non-pregnant (NP) control rats effect on cerebral haemodynamics and CBF (Belfort
despite similar pressures of autoregulatory breakthrough et al. 1999; Sherman et al. 2003; Hatab et al. 2005). For
(Euser & Cipolla, 2007). This suggests that pregnancy alone example, we previously showed that while magnesium

DOI: 10.1113/expphysiol.2007.039966 
C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
Exp Physiol 93.2 pp 254–261 Magnesium sulphate and blood–brain barrier permeability 255

sulphate has a modest vasodilatory effect on cerebral and animals treated with magnesium sulphate prior to
resistance arteries, the sensitivity of this response is acute hypertension (HTN + Mg2+ , n = 7). Sham rats did
decreased by pregnancy and the postpartum state (Euser & not undergo acute hypertension, though experimental
Cipolla, 2005). Furthermore, a randomized controlled trial length was comparable. For analysis of AQP4 protein
found that when compared with nimodipine, a calcium expression, separate groups of rats (n = 3 per group)
channel blocker with specific cerebral vasodilator action, were used: LP, LP + Mg2+ and non-pregnant (NP). These
magnesium sulphate was more effective in preventing animals did not undergo acute hypertension, since we were
eclamptic seizures (Belfort et al. 2003). Together, these interested in the effect of magnesium treatment on AQP4
results provide evidence that the primary action of protein expression only. All animals were housed in the
magnesium sulphate in eclampsia is likely not to be the University of Vermont Animal Care Facility, an American
relief of cerebral vasospasm. Association for the Accrediatation of Laboratory Animal
Treatment with magnesium sulphate has been reported Care accredited facility. All experimental procedures were
to decrease BBB permeability and cerebral oedema approved by the University of Vermont Institutional
formation in a variety of brain injury conditions, including Animal Care and Use Committee.
traumatic brain injury (Okiyama et al. 1995; Feldman
et al. 1996; Esen et al. 2003; Ghabriel et al. 2006), septic
encephalopathy (Esen et al. 2005), hypoglycaemia (Kaya In vivo model of HTE
et al. 2001) and hyperosmolar mannitol injection (Kaya
An in vivo model of HTE was used to determine
et al. 2004). We therefore hypothesized that the action
both BBB permeability and cerebral oedema formation
of magnesium sulphate in the prophylaxis of eclampsia
during acute hypertension, as previously described (Euser
may be related to protection of the BBB during acute
& Cipolla, 2007). This model of HTE is considered
hypertension. The goal of this study was to determine
a model of eclampsia (Cipolla, 2007) and has been
the effect of treatment with magnesium sulphate on in
used previously to study BBB permeability and oedema
vivo BBB permeability and formation of cerebral oedema
formation during pregnancy (Euser & Cipolla, 2007).
following CBF autoregulatory breakthrough in LP rats. In
Briefly, anaesthesia was initiated with isoflurane (≤ 3%
addition, it has been proposed that magnesium sulphate
in oxygen, inhaled; Abbott, North Chicago, IL, USA)
may limit cerebral oedema formation by decreasing the
and maintained with i.v. pentobarbital (≤ 60 mg kg−1 h−1 ;
expression of aquaporin-4 (AQP4; Ghabriel et al. 2006),
Ovation Pharmaceuticals, Deerfield, IL, USA), which was
a water channel protein highly expressed in the brain,
decreased during the surgical preparation, as tolerated,
although this interaction has not been directly shown.
to minimize the effects of anaesthesia on experimental
Therefore, another goal of this study was to determine
parameters. Adequate anaesthesia was assessed by toe
the effect of treatment with magnesium sulphate on AQP4
pinch and changes in arterial blood pressure. During
protein expression in LP rats without acute hypertension.
the course of each experiment, CBF was measured
transcranially using laser Doppler flowmetry with a
1.0 mm probe (Perimed, North Royalton, OH, USA)
Methods
affixed over a thinned area of skull (posterior to the coronal
Animals suture and lateral to the sagittal suture) over the middle
cerebral artery perfusion domain, as described elsewhere
All experiments used a rat model of pregnancy in (Smeda et al. 1999). All laser Doppler measurements
which primiparous Sprague–Dawley rats (Charles River, of CBF were normalized to the flow at baseline (after
St Constant, PQ, Canada) were studied on day 19–21 anaesthesia had been minimized and prior to acute
of a 22 day gestation. For each outcome measure, one hypertension) to determine a relative CBF (rCBF). The
group of animals was treated with an i.p. injection of following equation was used:
magnesium sulphate (270 mg kg−1 every 4 h for 24 h)
prior to acute hypertension. This dosage of magnesium rCBF = (CBFmax /CBFbaseline )
sulphate has been reported to produce serum magnesium
levels in rats within the range (4.2–8.4 mg dl−1 , 0.35– where CBFmax is the flow in laser Doppler units at maximal
0.70 mmol l−1 ) recommended for eclamptic seizure pressure and CBFbaseline is the flow in laser Doppler units
prophylaxis in pregnant women over 12–24 h (Pritchard, prior to acute hypertension and after anaesthesia had been
1955; Hallak et al. 1994; Leveno & Cunningham, 1999). For minimized. Therefore, a rCBF of 2.0 signifies a twofold
experiments investigating BBB permeability or cerebral increase in CBF from baseline.
oedema formation in vivo, three groups of LP rats Catheters were placed in the femoral artery and
were studied: animals without acute hypertension or veins for arterial blood pressure measurement and drug
magnesium sulphate treatment (sham, n = 10); animals delivery, respectively. Blood pressures were measured
that underwent acute hypertension alone (HTN, n = 7); via a pressure transducer attached in-line with the


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
256 A. G. Euser and others Exp Physiol 93.2 pp 254–261

catheter. Acute hypertension was induced by i.v. infusion quickly decapitated and the brain was then removed for
of phenylephrine (0.01 g per 10 ml lactated Ringer’s wet and dry weight measurements. The brain was weighed
solution; all reagents from Sigma, St Louis, MO, USA wet immediately after removal and then dried at 100◦ C
unless otherwise specified). Following 10 min of blood for 24 h, at which point the brain was weighed again dry.
pressure 180 mmHg, sufficient to cause autoregulatory Brain water content (as a percentage) was determined by
breakthrough (Euser & Cipolla, 2007), the experiment the following formula:
was ended with the humane death of the animal by
decapitation, and the brain was quickly removed. This Brain water content = [(weightwet − weightdry )
model of HTE was used with slight alterations to determine ÷ weightwet ] × 100%
both BBB permeability and cerebral oedema formation
during acute hypertension, described in the subsections where weightwet is the weight of the brain immediately after
below. removal from the animal and weightdry is the weight of the
brain after drying.
Blood–brain barrier permeability studies
Western blot analysis of AQP4 expression
To determine the extent of BBB permeability, a lactated
Ringer solution (100 ml of solution, USP contains: NaCl Following induction of anaesthesia with isoflurane
600 mg, NaC3 H5 O3 anhydrous 310 mg, KCl 30 mg and (Abbott, North Chicago, IL, USA) and decapitation,
CaCl2 dihydrate 20 mg. pH of the solution is 6.6 (6.0– brains were carefully removed and divided into
7.5).) Containing two different-sized dye tracers, 0.1% anterior and posterior cerebrum, as described above.
sodium fluorescein (NaFl, 376 Da) and 2% Evan’s Blue Brain sections were snap frozen in liquid nitrogen
(EB, 69 kDa), was infused into the left ventricle of the and stored at −80◦ C. For protein extraction, each
heart via catheter in the carotid artery. This solution was section was homogenized in a glass Dounce tissue
allowed to circulate for 10 min prior to induction of acute grinder with 3 ml lysis buffer comprising: 50 mm
hypertension by i.v. infusion of phenylephrine (0.01 g Trizma® hydrochloride, 150 mm NaCl, 10 mm EDTA,
per 10 ml lactated Ringer solution). Following 10 min 0.25% deoxycholate, 1% nonylphenyl polyethylene glycol
of blood pressure 180 mmHg, again sufficient to cause detergent (Calbiochem, San Diego, CA, USA), 10%
autoregulatory breakthrough (Euser & Cipolla, 2007), glycerol, 1% sodium dodecyl sulphate, 1 mm dithiothreitol
the animal was perfused with lactated Ringer solution (Bio-Rad, Richmond, CA, USA) and 1% protease inhibitor
through the central catheter in order to flush the dye cocktail. The homogenate was transferred and centrifuged
from the cerebral circulation. Following decapitation, the at 3900 g for 10 min at 4◦ C. The supernatant was
brain was quickly removed, sectioned and weighed. The centrifuged again under the same conditions. The total
cerebral cortices were separated from the cerebellum and amount of protein was measured using the Coomassie
brainstem, and then further divided into anterior and Plus-BradfordTM . Assay Kit (Pierce, Rockford, IL, USA).
posterior cerebrum sections by a coronal cut at the level Protein samples were incubated in Laemmli sample
of the optic chiasm. We chose to compare anterior and buffer (Bio-Rad) with 2β-mercaptoethanol at 95◦ C
posterior cerebrums because of the propensity of oedema for 10 min. Protein (10 µg) was separated by sodium
to form in the posterior brain regions during eclampsia dodecyl sulphate-polyacrylamide gel electrophoresis and
and PRES (Schwartz et al. 1992, 2000; Hinchey et al. 1996; transferred to a polyvinylidene difluoride membrane
Engelter et al. 2000; Lamy et al. 2004; Zeeman et al. 2004; (Bio-Rad). Membranes were blocked for 20 min at room
Cipolla, 2007). Any animal in which gross observation temperature in 3% non-fat milk in phosphate-buffered
revealed inadequate flushing of the cerebrovasculature saline containing 0.005% Tween-20 (PBST; Calbiochem),
was excluded from analysis (this occurred in 2 animals). cut vertically, and subsequently incubated overnight at
Samples were processed as previously described (Euser & 4◦ C with two primary antibodies: an affinity purified
Cipolla, 2007), and the resulting supernatant was analysed rabbit polyclonal antibody raised against residues 249–
by fluorescence spectrophotometry at 460–515 nm for 323 of rat aquaporin-4, 1:1000 (Chemicon, Temecula,
NaFl and 620–680 nm for EB. Data are expressed as average CA, USA), and a mouse monoclonal antibody to
fluorescence counts per second (CPS) per gram brain glyceraldehyde-3-phosphate dehydrogenase (GAPDH),
tissue. 1:30 000 (Biodesign, Saco, ME, USA). After repeated
washing in PBST, membranes were incubated in secondary
antibodies conjugated to horseradish peroxidase for 1 h
Determination of cerebral oedema
at room temperature. A sheep antirabbit IgG (Abcam,
The percentage water content of the brain is a measure of Cambridge, MA, USA), 1:2000, was used for AQP4
cerebral oedema (Schwab et al. 1997). After elevated blood and a goat antimouse IgG, 1:3000 (Pierce), was used
pressure had been maintained for 10 min, animals were for GAPDH. Additional washing steps in PBST were


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
Exp Physiol 93.2 pp 254–261 Magnesium sulphate and blood–brain barrier permeability 257

Table 1. Physiological characteristics of animals studied


Weight (g) Litter size Baseline ABP (mmHg) Maximal ABP (mmHg) Maximal rCBF
Sham (n = 10) 344 ± 4 13 ± 1 118 ± 5 126 ± 5 1.07 ± 0.027
HTN (n = 7) 336 ± 11 12 ± 1 116 ± 4 191 ± 3∗∗ 2.41 ± 0.29∗∗
HTN + Mg2+ (n = 7) 342 ± 15 12 ± 1 102 ± 3† 186 ± 4∗∗ 2.21 ± 0.30∗∗
Abbreviations: ABP, arterial blood pressure; rCBF, relative cerebral blood flow; and HTN, acute hypertension. ∗∗ P < 0.01 versus
sham group within same experimental conditions; †P < 0.05 versus sham and HTN.

followed by chemiluminescence using SuperSignal West t test. Likewise, differences between baseline and maximal
Pico Substrate and CL-XPosure Film (Pierce). Films were blood pressures within the same treatment group were
scanned into Adobe Photoshop CS, and densitometry determined by Student’s paired t test. Differences in AQP4
of AQP4 and GAPDH bands was determined with expression between LP and LP + Mg2+ groups were tested
ImageJ software (version 1.37v, available for download by non-parametric Mann–Whitney U test. Differences
at http://rsb.info.nih.gov/ij/). All experiments were done were considered significant if P < 0.05.
in duplicate. The ratio of AQP4/GAPDH intensity was
calculated for each animal in each group. Values for NP
Results
animals were assigned a value of 1.0 and LP and LP + Mg2+
values were normalized to the NP group before analysing The physiological characteristics of the LP animals studied
each group. with the in vivo model of HTE are summarized in
Table 1. Two animals were excluded from study, one from
each group of HTN and HTN + Mg2+ animals, owing
Statistical analyses to insufficient flushing of the dye from the vasculature.
All data are expressed as means ± s.e.m. Differences in There were no significant differences between treatment
animal characteristics, tissue fluorescence and brain water groups in either animal weight or litter size. Baseline
content between treatment groups were determined by mean arterial blood pressure, measured directly by femoral
ANOVA with a post hoc Student–Newman–Keuls test for arterial catheter, was not significantly different between
multiple comparisons. Differences in tissue fluorescence sham and HTN groups, but was significantly lower in
between the anterior and posterior cerebrum within the animals treated with magnesium (P < 0.05). Infusion
same treatment group were determined by Student’s paired of phenylephrine to induce autoregulatory breakthrough
caused a significant increase in arterial blood pressure in
the HTN and HTN + Mg2+ groups compared with the
5000 sham group (P < 0.01). The increase in blood pressure
Sham
was accompanied by a significant increase in CBF, as
4000
HTN represented by the rCBF ≥ 2.0 for both hypertensive
HTN + Mg *‡ groups, indicating autoregulaotry breakthrough (P < 0.01
Fluorescence (CPS/g)

versus sham).
3000
Acute hypertension with autoregulatory breakthrough
* *† significantly increased BBB permeability to EB in both
2000 the anterior and posterior cerebrum (Fig. 1). Interestingly,
the increase in BBB permeability was significantly greater
1000 in the posterior brain region versus anterior (659 versus
365%, respectively, P < 0.05), a region most susceptible
to oedema formation. Magnesium sulphate treatment
0
Anterior Posterior
Cerebrum
decreased BBB permeability significantly only in the
Cerebrum
Brain Region posterior brain region (Fig. 1). There was a 41% decrease
in permeability in the posterior cerebrum (P < 0.05). The
Figure 1. Bar graph showing average fluorescence (in CPS g−1 ) BBB permeability to NaFl was unaffected by hypertension
of Evan’s Blue in the anterior and posterior cerebrum of all
groups of late-pregnant animals as a measure of BBB
or magnesium sulphate treatment (Fig. 2). The lack of a
permeability significant difference in NaFl permeability probably results
Blood–brain barrier permeability was significantly increased with acute from the higher variability in those groups compared with
hypertension and autoregulatory breakthrough (HTN) compared with EB.

sham control animals ( P < 0.05). The increase in BBB permeability Brain water content, a measure of cerebral oedema
was greater in the posterior versus anterior cerebrum (‡P < 0.05).
Magnesium sulphate treatment (HTN + Mg2+ ) significantly decreased
formation (Schwab et al. 1997), was not different between
BBB permeability in the posterior cerebrum in response to acute any of the groups. The percentage water content was:
hypertension (†P < 0.05 versus HTN). 78.25 ± 0.18% for sham group, 78.13 ± 0.02% for LP


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
258 A. G. Euser and others Exp Physiol 93.2 pp 254–261

50 Sham HTN group and 78.02 ± 0.07% for LP HTN + Mg2+


HTN group (n.s.). In contrast to our previous findings (Euser
HTN + Mg & Cipolla, 2007), there was no significant difference in
40
Fluorescence (CPS/g) x 1,000

cerebral oedema formation between any of the groups.


Aquaporin-4 protein expression was determined in
30 the anterior and posterior cerebrum of normotensive LP
and LP + Mg2+ groups and compared with NP control
20 animals. Expression of AQP4 was increased in LP and
LP + Mg2+ compared with NP animals in both the
anterior and posterior cerebrum by 3- to 4-fold (Fig. 3),
10
a finding similar to previous results (Quick & Cipolla,
2005). Contrary to previous reports (Ghabriel et al. 2006),
0 magnesium sulphate treatment did not have any effect on
Anterior Posterior
Cerebrum Cerebrum AQP4 expression.
Brain Region

Figure 2. Bar graph showing average fluorescence (in CPS g−1 ) Discussion
of sodium fluorescein in the anterior and posterior cerebrum in
all groups of late-pregnant rats as a measure of BBB The major finding of this study was that magnesium
permeability sulphate treatment significantly decreased BBB
There was no difference in BBB between animals that underwent
acute hypertension (HTN) compared with sham control animals.
permeability to EB in LP rats during acute hypertension,
Magnesium sulphate treatment (HTN + Mg2+ ) also had no effect on an effect that was significant in the posterior brain
BBB permeability to NaFl compared with HTN. region. In fact, BBB permeability to EB varied regionally

Figure 3. Aquaporin-4 (AQP4) expression in the female rat brain with and without magnesium
treatment
Representative Western blots of AQP4 expression in late-pregnant (LP) rats in the anterior cerebrum (A) and
posterior cerebrum (B). Glyceraldehyde-3-phosphate dehydrogenase (GAPDH) blots from the same gel were used
as an internal control. C shows the intensity ratios for untreated (LP) and treated animals (LP + Mg2+ ) compared
with non-pregnant control animals (NP). Aquaporin-4 expression was increased in both LP and LP + Mg2+ animals
compared with NP control animals, but was unchanged by magnesium sulphate treatment.


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
Exp Physiol 93.2 pp 254–261 Magnesium sulphate and blood–brain barrier permeability 259

in response to autoregulatory breakthrough, such that less in the present study compared with what we have
the posterior cerebrum showed a greater increase in published previously. The purpose of the previous study
permeability than the anterior cerebrum. Since the (Euser & Cipolla, 2007) was to determine the pressure
posterior brain region is most susceptible to oedema at which autoregulatory breakthrough occurred between
formation during PRES and eclampsia (Schwartz et al. NP and LP rats, and those animals routinely had rCBF
1992, 2000; Hinchey et al. 1996; Engelter et al. 2000; > 3.0. This difference would make hydrostatic pressure
Lamy et al. 2004; Zeeman et al. 2004; Cipolla, 2007), these less in the present group of animals compared with
results suggest that one mechanism by which oedema the previous study. Hydrostatic pressure is a primary
forms preferentially in this region is the enhanced BBB determinant of brain oedema (Shima, 2003). Therefore,
permeability to large proteins such as EB. Together, these a lower rCBF would suggest a lower hydrostatic pressure
results suggest that the beneficial effect of magnesium and could account for the undetectable changes in brain
sulphate treatment during eclampsia may be related to water content in the present study. It is also possible that
protection of the BBB that limits solute flux into the the duration of acute hypertension, being only 10 min,
brain tissue when hydrostatic pressure is elevated to was not sufficient to cause a detectable increase in oedema
pathological levels. formation with this degree of rCBF change. While this
The mechanism by which magnesium sulphate acts methodology has been commonly used to study BBB
to decrease BBB permeability is not clear from this permeability in response to acute hypertension, it probably
study, but it may be related to a direct effect on the takes some time for oedema to form. Unfortunately, this
cerebral endothelium. Magnesium sulphate is a calcium non-survival model of HTE does not allow pressure to be
antagonist (Fawcett et al. 1999) and may decrease increased for longer durations (probably due to peripheral
paracellular transport through tight junctions by opposing vascular dilatation that decreases total peripheral vascular
calcium-induced contractions of the actin cytoskeleton resistance and drops arterial blood pressure). Other
in endothelial cells. Alternatively, acute hypertension has methods in which the animals survive for longer periods,
been shown to increase pinocytosis that may enhance such as 2–3 days, would be better for assessment of
transcellular transport in the cerebral endothelium (Nag oedema in response to acute hypertension and magnesium
et al. 1977, 1979; Westergaard et al. 1977). Magnesium sulphate treatment.
may counter this mode of transport and decrease BBB Previous studies have suggested that magnesium
permeability during acute hypertension. In fact, it has sulphate treatment may limit cerebral oedema formation
been suggested that transport of large molecules across the by decreasing the expression of AQP4 (Ghabriel et al.
BBB implicates transcellular versus paracellular transport 2006). Aquaporin-4 is a transmembrane water channel
(Mayhan & Heistad, 1985). Since magnesium treatment highly expressed in the brain and shown to be upregulated
had a greater effect on a large solute (EB) than on a small during pregnancy (Quick & Cipolla, 2005). Owing
solute (NaFl), this suggests that transcellular transport to its water fluxing capabilities (Papadopoulos et al.
was the primary mode of BBB permeability under these 2004), a decrease induced by magnesium may limit
conditions. oedema formation. For this reason, we investigated
Another protective effect of magnesium sulphate on whether magnesium sulphate treatment affected AQP4
limiting BBB permeability during acute hypertension may protein expression. Animals were treated with magnesium
be related to its ability to scavenge free radicals. Free sulphate for 24 h, a duration consistent with clinical
radical production during acute hypertension induced by practice, but perhaps not long enough to have an effect
pressor agents has been demonstrated (Zhang & Ellis, on AQP4 protein levels. Figure 3 shows that AQP4 was
1991). In fact, superoxide dismutase, an oxygen free radical increased in both pregnant groups compared with NP
scavenger, was shown to reduce BBB permeability and animals, but magnesium sulphate treatment had no effect
cerebral oedema during acute hypertension in rats (Zhang on its expression. These measurements were done on
& Ellis, 1990). It is therefore possible that magnesium normotensive animals because we wanted to determine
sulphate acts similarly to scavenge free radicals and protect whether there was any effect of magnesium treatment prior
the BBB, since treatment with magnesium sulphate has to a hypertensive insult that may limit oedema formation.
been shown to decrease lipid peroxidation in pre-eclamptic How AQP4 protein expression changes in response to
women, suggesting that it has free radical scavenging hypertension is of clear interest; however, animals would
properties (Ariza et al. 2005). probably need to survive for longer time periods.
In contrast to a previous study (Euser & Cipolla, 2007), This study used a model of HTE during pregnancy
we did not find that cerebral oedema formation was to investigate how acute hypertension affects BBB
increased in response to acute hypertension in LP rats, or permeability. This model has been extensively used
that magnesium sulphate treatment affected brain water to investigate the effect of cerebral haemodynamics
content. This negative and contrary finding may be for on BBB permeability and vasogenic oedema formation
several reasons. First, the change in rCBF was considerably (MacKenzie et al. 1976; Baumbach & Heistad, 1985;


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
260 A. G. Euser and others Exp Physiol 93.2 pp 254–261

Hatashita et al. 1985, 1986; Mayhan & Heistad, 1985; Cipolla MJ (2007). Cerebrovascular changes in pregnancy and
Mayhan et al. 1986; Euser & Cipolla, 2007) and is eclampsia. Hypertension 50, 14–24.
considered a model of eclampsia (Cipolla, 2007). It is worth Easton JD (1998). Severe preeclampsia/eclampsia: hypertensive
noting, however, that we studied the effect of magnesium encephalopathy of pregnancy? Cerebrovasc Dis 8, 53–58.
treatment on BBB permeability during normal pregnancy, Engelter ST, Provenzale JM & Petrella JR (2000). Assessment of
vasogenic edema in eclampsia using diffusion imaging.
not under conditions of oxidative stress or endothelial cell
Neuroradiology 42, 818–820.
damage which may have a role in eclampsia. However, Esen F, Erdem T, Aktan D, Kalayci R, Cakar N, Kaya M & Telci
this model is valuable and has a number of the features L (2003). Effects of magnesium administration on brain
relevant to the pathogenesis of eclampsia, including loss edema and blood–brain barrier breakdown after
of autoregulation, hyperperfusion and BBB disruption. It experimental traumatic brain injury in rats. J Neurosurg
is also useful for evaluating regional differences in BBB Anesthesiol 15, 119–125.
permeability, which are an important feature considering Esen F, Erdem T, Aktan D, Orhan M, Kaya M, Eraksoy H,
the regional heterogeneity in oedema formation during Cakar N & Telci L (2005). Effect of magnesium sulfate
eclampsia. administration on blood–brain barrier in a rat model of
To our knowledge, this is the first study to investigate intraperitoneal sepsis: a randomized controlled experimental
the effect of magnesium sulphate treatment on BBB study. Crit Care 9, R18–R23.
Euser AG & Cipolla MJ (2005). Resistance artery vasodilation
permeability during acute hypertension in pregnancy.
to magnesium sulfate during pregnancy and the postpartum
The results demonstrate that treatment with magnesium state. Am J Physiol Heart Circ Physiol 288, H1521–H1525.
sulphate decreased BBB permeability following acute Euser AG & Cipolla MJ (2007). Cerebral blood flow
hypertension, particularly in the posterior cerebrum. autoregulation and edema formation during pregnancy in
However, magnesium treatment did not affect cerebral anesthetized rats. Hypertension 49, 334–340.
oedema formation during the short time course studied. A Fawcett WJ, Haxby EJ & Male DA (1999). Magnesium:
more complete understanding of the effect of magnesium physiology and pharmacology. Br J Anaesth 83, 302–320.
sulphate on the BBB may provide information regarding Feldman Z, Gurevitch B, Artru AA, Oppenheim A, Shohami E,
the beneficial effect of magnesium in the treatment and Reichenthal E & Shapira Y (1996). Effect of magnesium
prophylaxis of eclampsia. given 1 hour after head trauma on brain edema and
neurological outcome. J Neurosurg 85, 131–137.
Ghabriel MN, Thomas A & Vink R (2006). Magnesium restores
References altered aquaporin-4 immunoreactivity following traumatic
brain injury to a pre-injury state. Acta Neurochir Suppl 96,
Altman D, Carroli G, Duley L, Farrell B, Moodley J, Neilson J, 402–406.
Smith D & Magpie Trial Collaboration Group (2002). Do Hallak M, Berman RF, Irtenkauf SM, Janusz C & Cotton DB
women with pre-eclampsia, and their babies, benefit from (1994). Magnesium sulfate treatment decreases N -methyl-
magnesium sulphate? The Magpie Trial: a randomised D-aspartate receptor binding in the rat brain: an
placebo-controlled trial. Lancet 359, 1877–1890. autoradiographic study. J Soc Gynecol Invest 1, 25–30.
Ariza AC, Bobadilla N, Fernandez C, Munoz-Fuentes RM, Hatab MR, Zeeman GG & Twickler DM (2005). The effect
Larrea F & Halhali A (2005). Effects of magnesium sulfate on of magnesium sulfate on large cerebral artery blood flow
lipid peroxidation and blood pressure regulators in in severe preeclampsia. J Matern Fetal Neonat Med 17,
preeclampsia. Clin Biochem 38, 128–133. 187–192.
Baumbach GL & Heistad DD (1985). Heterogeneity of brain Hatashita S, Hoff JT & Ishii S (1986). Focal brain edema
blood flow and permeability during acute hypertension. associated with acute arterial hypertension. J Neurosurg 64,
Am J Physiol Heart Circ Physiol 249, H629–H637. 643–649.
Belfort MA, Anthony J, Saade GR, Allen JC Jr & Nimodipine Hatashita S, Koike J, Sonokawa T & Ishii S (1985). Cerebral
Study Group (2003). A comparison of magnesium sulfate edema associated with craniectomy and arterial
and nimodipine for the prevention of eclampsia. hypertension. Stroke 16, 661–668.
N Engl J Med 348, 304–311. Hinchey J, Chaves C, Appignani B, Breen J, Pao L, Wang A,
Belfort MA & Moise KJ Jr (1992). Effect of magnesium sulfate Pessin MS, Lamy C, Mas JL & Caplan LR (1996). A reversible
on maternal brain blood flow in preeclampsia: a randomized, posterior leukoencephalopathy syndrome. N Engl J Med 334,
placebo-controlled study. Am J Obstet Gynecol 167, 661–666. 494–500.
Belfort MA, Saade GR & Moise KJ Jr (1993). The effect of Kaya M, Gulturk S, Elmas I, Arican N, Kocyildiz ZC, Kucuk M,
magnesium sulfate on maternal and fetal blood flow in Yorulmaz H & Sivas A (2004). The effects of magnesium
pregnancy-induced hypertension. Acta Obstet Gynecol Scand sulfate on blood–brain barrier disruption caused by
72, 526–530. intracarotid injection of hyperosmolar mannitol in rats. Life
Belfort MA, Saade GR, Yared M, Grunewald C, Herd JA, Varner Sci 76, 201–212.
MA & Nisell H (1999). Change in estimated cerebral Kaya M, Kucuk M, Kalayci RB, Cimen V, Gurses C, Elmas I &
perfusion pressure after treatment with nimodipine or Arican N (2001). Magnesium sulfate attenuates increased
magnesium sulfate in patients with preeclampsia. blood–brain barrier permeability during insulin-induced
Am J Obstet Gynecol 181, 402–407. hypoglycemia in rats. Can J Physiol Pharmacol 79, 793–798.


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society
Exp Physiol 93.2 pp 254–261 Magnesium sulphate and blood–brain barrier permeability 261

Lamy C, Oppenheim C, Meder JF & Mas JL (2004). Schwartz RB, Jones KM, Kalina P, Bajakian RL, Mantello
Neuroimaging in posterior reversible encephalopathy MT, Garada B & Holman BL (1992). Hypertensive
syndrome. J Neuroimaging 14, 89–96. encephalopathy: findings on CT, MR imaging, and
Leveno KJ & Cunningham FG (1999). Management of SPECT imaging in 14 cases. AJR Am J Roentgenol 159,
preeclampsia. In Chesley’s Hypertensive Disorders in 379–383.
Pregnancy, ed. Lindheimer MD, Roberts JM & Cunningham Sherman R, Armory P, Moody P, Hope T & Mahajan RP
FG, pp. 543–580. Appleton & Lange, Stamford, CT, USA. (2003). Effects of magnesium sulphate on cerebral
MacKenzie ET, Strandgaard S, Graham DI, Jones JV & Harper haemodynamics in healthy volunteers: a transcranial
AM (1976). Effects of acutely induced hypertension in cats Doppler study. Br J Anaesth 91, 273–275.
on pial arteriolar caliber, local cerebral blood flow, and the Shima K (2003). Hydrostatic brain edema: basic mechanisms
blood–brain barrier. Circ Res 39, 33–41. and clinical aspect. Acta Neurochir Suppl 86, 17–20.
Mayhan WG, Faraci FM & Heistad DD (1986). Disruption of Smeda JS, VanVliet BN & King SR (1999). Stroke-prone
the blood–brain barrier in cerebrum and brain stem during spontaneously hypertensive rats lose their ability to
acute hypertension. Am J Physiol Heart Circ Physiol 251, auto-regulate cerebral blood flow prior to stroke. J Hypertens
H1171–H1175. 17, 1697–1705.
Mayhan WG & Heistad DD (1985). Permeability of blood– The Eclampsia Trial Collaborative Group(1995). Which
brain barrier to various sized molecules. Am J Physiol Heart anticonvulsant for women with eclampsia? Evidence from
Circ Physiol 248, H712–H718. the Collaborative Eclampsia Trial. Lancet 345, 1455–1463.
Nag S, Robertson DM & Dinsdale HB (1977). Cerebral cortical Westergaard E, van Deurs B & Bronsted HE (1977). Increased
changes in acute experimental hypertension: an vesicular transfer of horseradish peroxidase across cerebral
ultrastructural study. Lab Invest 36, 150–161. endothelium, evoked by acute hypertension. Acta
Nag S, Robertson DM & Dinsdale HB (1979). Quantitative Neuropathol (Berl) 37, 141–152.
estimate of pinocytosis in experimental acute hypertension. Witlin AG & Sibai BM (1998). Magnesium sulfate therapy in
Acta Neuropathol (Berl) 46, 107–116. preeclampsia and eclampsia. Obstet Gynecol 92, 883–889.
Naidu S, Payne AJ, Moodley J, Hoffmann M & Gouws E (1996). Zeeman GG, Fleckenstein JL, Twickler DM & Cunningham FG
Randomised study assessing the effect of phenytoin and (2004). Cerebral infarction in eclampsia. Am J Obstet Gynecol
magnesium sulphate on maternal cerebral circulation in 190, 714–720.
eclampsia using transcranial Doppler ultrasound. Br J Obstet Zhang XM & Ellis EF (1990). Superoxide dismutase reduces
Gynaecol 103, 111–116. permeability and edema induced by hypertension in rats.
Okiyama K, Smith DH, Gennarelli TA, Simon RP, Leach M & Am J Physiol Heart Circ Physiol 259, H497–H503.
McIntosh TK (1995). The sodium channel blocker and Zhang XM & Ellis EF (1991). Superoxide dismutase decreases
glutamate release inhibitor BW1003C87 and magnesium mortality, blood pressure, and cerebral blood flow responses
attenuate regional cerebral edema following experimental induced by acute hypertension in rats. Stroke 22, 489–494.
brain injury in the rat. J Neurochem 64, 802–809.
Papadopoulos MC, Manley GT, Krishna S & Verkman AS
(2004). Aquaporin-4 facilitates reabsorption of excess fluid
in vasogenic brain edema. FASEB J 18, 1291–1293.
Pritchard JA (1955). The use of the magnesium ion in the
management of eclamptogenic toxemias. Surg Gynecol Obstet
100, 131–140.
Quick AM & Cipolla MJ (2005). Pregnancy-induced
up-regulation of aquaporin-4 protein in brain and its role in Acknowledgements
eclampsia. FASEB J 19, 170–175.
Schwab M, Bauer R & Zwiener U (1997). The distribution of We gratefully acknowledge the support of the American
normal brain water content in Wistar rats and its increase Heart Association Established Investigator Award (0540081N
due to ischemia. Brain Res 749, 82–87. to M.J.C.), the American Heart Association North-east Affiliate
Schwartz RB, Feske SK, Polak JF, DeGirolami U, Iaia A, Beckner Research Committee Predoctoral Fellowship (000019871 to
KM, Bravo SM, Klufas RA, Chai RY & Repke JT (2000). A.G.E.), the National Institute of Neurological Disorders and
Preeclampsia-eclampsia: clinical and neuroradiographic Stroke (R01 NS-045940 to M.J.C.), the Totman Medical Research
correlates and insights into the pathogenesis of hypertensive Trust, and the University of Vermont College of Medicine
encephalopathy. Radiology 217, 371–376. MD/PhD Program.


C 2007 The Authors. Journal compilation 
C 2007 The Physiological Society

You might also like